US20150231219A1 - Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies - Google Patents

Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies Download PDF

Info

Publication number
US20150231219A1
US20150231219A1 US14/388,330 US201314388330A US2015231219A1 US 20150231219 A1 US20150231219 A1 US 20150231219A1 US 201314388330 A US201314388330 A US 201314388330A US 2015231219 A1 US2015231219 A1 US 2015231219A1
Authority
US
United States
Prior art keywords
cancer
igf
inhibitor
administration
bispecific
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/388,330
Other languages
English (en)
Inventor
Alexey Alexandrovich Lugovskoy
Jason Baum
Sharlene Adams
Bryan Johnson
Jian Tang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merrimack Pharmaceuticals Inc
Original Assignee
Merrimack Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merrimack Pharmaceuticals Inc filed Critical Merrimack Pharmaceuticals Inc
Priority to US14/388,330 priority Critical patent/US20150231219A1/en
Assigned to MERRIMACK PHARMACEUTICALS, INC. reassignment MERRIMACK PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ADAMS, SHARLENE, BAUM, Jason, JOHNSON, BRYAN, LUGOVSKOY, ALEXEY ALEXANDROVICH
Assigned to MERRIMACK PHARMACEUTICALS, INC. reassignment MERRIMACK PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ADAMS, SHARLENE, BAUM, Jason, LUGOVSKOY, ALEXEY ALEXANDROVICH
Assigned to MERRIMACK PHARMACEUTICALS, INC. reassignment MERRIMACK PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TANG, JIAN
Publication of US20150231219A1 publication Critical patent/US20150231219A1/en
Assigned to U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT reassignment U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MERRIMACK PHARMACEUTICALS, INC.
Assigned to MERRIMACK PHARMACEUTICALS, INC. reassignment MERRIMACK PHARMACEUTICALS, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: U.S. BANK NATIONAL ASSOCIATION
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/568Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone
    • A61K31/5685Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone having an oxo group in position 17, e.g. androsterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001106Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/852Pancreas
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific

Definitions

  • Tumor cells express receptors for growth factors and cytokines that stimulate proliferation of the cells. Antibodies to such receptors can be effective in blocking the stimulation of cell proliferation mediated by growth factors and cytokines and can thereby inhibit tumor cell proliferation and tumor growth.
  • Commercially available therapeutic antibodies that target receptors on cancer cells include, for example, trastuzumab which targets the HER2 receptor (also known as ErbB2) for the treatment of breast cancer, and cetuximab which targets the epidermal growth factor receptor (EGFR, also known as HER1 or ErbB1) for the treatment of colorectal cancer and head and neck cancer.
  • trastuzumab which targets the HER2 receptor (also known as ErbB2) for the treatment of breast cancer
  • cetuximab which targets the epidermal growth factor receptor (EGFR, also known as HER1 or ErbB1) for the treatment of colorectal cancer and head and neck cancer.
  • EGFR epidermal growth factor receptor
  • Monoclonal antibodies have significantly advanced our ability to treat cancers, yet clinical studies have shown that many patients do not adequately respond to monospecific therapy. This is in part due to the multigenic nature of cancers, where cancer cells rely on multiple and often redundant pathways for proliferation. Bi- or multi-specific antibodies capable of blocking multiple growth and survival pathways at once have a potential to better meet the challenge of blocking cancer growth, and indeed many of them are advancing in clinical development.
  • the co-administration of pluralities of anti-cancer drugs often provides better treatment outcomes than monotherapy.
  • PBA polyvalent bispecific antibodies
  • Monotherapy with a bispecific anti-IGF-1R and anti-ErbB3 antibody suppresses tumor growth in a dose-dependent manner in in vivo xenograft models of a variety of cancers including pancreatic cancer, renal cell carcinoma, Ewing's sarcoma, non-small cell lung cancer, gastrointestinal neuroendocrine cancer, estrogen receptor positive locally advanced or metastatic cancer, ovarian cancer, colorectal cancer, endometrial cancer, or glioblastoma.
  • additional anti-cancer agents such as everolimus, capecitabine, or XL147, exhibits therapeutic synergy.
  • kits for the treatment of a cancer in a human patient by administering an effective amount of a bispecific anti-IGF-1R and anti-ErbB3 antibody to the patient, where the patient is given a single loading dose of at least 10 mg/kg of the bispecific antibody followed administration of one or more maintenance doses given at intervals.
  • the intervals between doses are intervals of at least three days. In some embodiments, the intervals are every seven days, every fourteen days or every twenty-one days.
  • the doses administered may range from 1 mg/kg to 60 mg/kg of the bispecific antibody. In some embodiments, the loading dose is greater than the maintenance dose.
  • the loading dose may from 12 mg/kg to 20 mg/kg, from 20 mg/kg to 40 mg/kg, or from 40 mg/kg to 60 mg/kg. In some embodiments the loading dose is about 12 mg/kg, 20 mg/kg, 40 mg/kg, or 60 mg/kg. In other embodiments the maintenance dose is about 6 mg/kg, 12 mg/kg, 20 mg/kg, 30 mg/kg, 40 mg/kg, 50 mg/kg or 60 mg/kg.
  • the patient has a pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, Ewing's sarcoma, non-small cell lung cancer, gastrointestinal neuroendocrine cancer, estrogen receptor- or progesterone receptor-positive locally advanced or metastatic breast cancer, ovarian cancer, triple negative breast cancer, colorectal cancer, endometrial cancer, or glioblastoma.
  • the patient has a cancer that is refractory to one or more anti-cancer agents, e.g., gemcitabine or sunitinib.
  • the bispecific anti-IGF-1R and anti-ErbB3 antibody has an anti-IGF-1R module selected from the group consisting of SF, P4, M78, and M57. In another embodiment the bispecific anti-IGF-1R and anti-ErbB3 antibody has an anti-ErbB3 module selected from the group consisting of C8, P1, M1.3, M27, P6, and B69. In one embodiment, the bispecific anti-IGF-1R and anti-ErbB3 antibody is P4-G1-M1.3. In another embodiment, the bispecific anti-IGF-1R and anti-ErbB3 antibody is P4-G1-C8.
  • Also provided are methods of providing treatment of cancer in a human patient comprising co-administering to the patient an effective amount each of a bispecific anti-IGF-1R and anti-ErbB3 antibody and of one or more additional anti-cancer agents, wherein the anti-cancer agent is a PI3K pathway inhibitor, an mTOR inhibitor, a MEK inhibitor, a multikinase inhibitor, a B-Raf inhibitor, a taxane, irinotecan, nanoliposomal irinotecan, an anti-endocrine therapy, an antihormonal therapy, or an antimetabolite therapy.
  • the anti-cancer agent is an mTOR inhibitor.
  • Exemplary mTOR inhibitors are selected from the group comprising everolimus, temsirolimus, sirolimus, or ridaforolimus.
  • the mTOR inhibitor is a pan-mTOR inhibitor selected from the group consisting of INK128, CC223, OSI207, AZD8055, AZD2014, and Palomid529.
  • the anti-cancer agent is a phosphoinositide-3-kinase (PI3K) inhibitor or PI3K pathway inhibitor, e.g., perifosine (KRX-0401), SF1126, CAL101, BKM120, BKM120, XL147, or PX-866.
  • PI3K phosphoinositide-3-kinase
  • PI3K pathway inhibitor e.g., perifosine (KRX-0401), SF1126, CAL101, BKM120, BKM120, XL147, or PX-866.
  • the PI3K inhibitor is XL147 or BKM120.
  • the anti-cancer agent is a MEK inhibitor, e.g., GSK1120212.
  • the anti-cancer agent is a multikinase inhibitor.
  • the multikinase inhibitor is sorafenib.
  • the anti-cancer agent is an antimetabolite therapy, e.g., gemcitabine, capecitabine, cytarabine, or 5-fluorouracil.
  • the antimetabolite is gemcitabine.
  • the antimetabolite is a taxane such as docetaxel, cabazitaxel, nab-paclitaxel, or paclitaxel. In another embodiment, the antimetabolite is capecitabine or 5-fluorouracil. In some embodiments, the anti-cancer agent is irinotecan or nanoliposomal irinotecan. In another embodiment, the anti-cancer agent is a B-Raf inhibitor. In some embodiments, the anti-cancer agent is antihormonal therapy. In certain embodiments, then antihormonal therapy is tamoxifen, exemestane, letrozole, or fulvestrant.
  • co-administration of the additional anti-cancer agent or agents has an additive or superadditive effect on suppressing tumor growth, as compared to administration of the bispecific anti-IGF-1R and anti-ErbB3 antibody alone or the one or more additional anti-cancer agents alone, wherein the effect on suppressing tumor growth is measured in a mouse xenograft model using BxPC-3, Caki-1, SK-ES-1, A549, NCI/ADR-RES, BT-474, DU145, or MCF7 cells.
  • compositions for use in the treatment of a cancer, or for the manufacture of a medicament for the treatment of cancer comprising a bispecific anti-IGF-1R and anti-ErbB3 antibody to be administered to a patient requiring treatment of a cancer, the administration comprising administering to the patient a single loading dose of at least 10 mg/kg of the bispecific antibody followed by administration of one or more maintenance doses given at intervals.
  • the intervals between doses are intervals of at least three days. In some embodiments, the intervals between doses are every fourteen days or every twenty-one days.
  • the compositions comprise a loading dose that is greater than the maintenance dose.
  • the loading dose may from about 12 mg/kg to about 20 mg/kg, from about 20 mg/kg to about 40 mg/kg, or from about 40 mg/kg to about 60 mg/kg.
  • the loading dose is about 12 mg/kg, about 20 mg/kg, about 40 mg/kg, or about 60 mg/kg.
  • the maintenance dose is about 6 mg/kg, about 12 mg/kg, about 20 mg/kg, about 30 mg/kg, about 40 mg/kg, about 50 mg/kg or about 60 mg/kg.
  • the patient has a cancer that is refractory to one or more anti-cancer agents, e.g., gemcitabine, sunitinib, or sorafenib.
  • the patient has a pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, Ewing's sarcoma, non-small cell lung cancer, gastrointestinal neuroendocrine cancer, estrogen receptor-positive locally advanced or metastatic cancer, ovarian cancer, colorectal cancer, endometrial cancer, or glioblastoma.
  • the bispecific anti-IGF-1R and anti-ErbB3 antibody has an anti-IGF-1R module selected from the group consisting of SF, P4, M78, and M57. In another embodiment the bispecific anti-IGF-1R and anti-ErbB3 antibody has an anti-ErbB3 module selected from the group consisting of C8, P1, M1.3, M27, P6, and B69. In one embodiment, the bispecific anti-IGF-1R and anti-ErbB3 antibody is P4-G1-M1.3. In another embodiment, the bispecific anti-IGF-1R and anti-ErbB3 antibody is P4-G1-C8.
  • compositions comprise an effective amount each of a bispecific anti-IGF-1R and anti-ErbB3 antibody and of one or more additional anti-cancer agents, wherein the anti-cancer agent is a PI3K pathway inhibitor, an mTOR inhibitor, a MEK inhibitor, a multikinase inhibitor, a B-Raf inhibitor, nanoliposomal irinotecan, or an antimetabolite.
  • the anti-cancer agent is an mTOR inhibitor.
  • the mTOR inhibitor is selected from the group comprising everolimus, temsirolimus, sirolimus, or ridaforolimus.
  • the mTOR inhibitor is a pan-mTOR inhibitor chosen from the group consisting of INK128, CC223, OSI207, AZD8055, AZD2014, and Palomid529.
  • the anti-cancer agent is a phosphoinositide-3-kinase (PI3K) inhibitor, e.g., perifosine (KRX-0401), SF1126, CAL101, BKM120, BKM120, XL147, or PX-866.
  • the PI3K inhibitor is XL147.
  • the anti-cancer agent is a MEK inhibitor.
  • Exemplary MEK inhibitors are selected from the group consisting of GSK1120212, BAY 86-9766, or AZD6244.
  • the anti-cancer agent is a multikinase inhibitor.
  • the multikinase inhibitor is sorafenib or sunitinub.
  • the anti-cancer agent is an antimetabolite, e.g., gemcitabine, docetaxel, paclitaxel, capecitabine, cytarabine, or 5-fluorouracil.
  • the anti-cancer agent is nanoliposomal irinotecan.
  • the anti-cancer agent is a B-Raf inhibitor.
  • the composition comprises a bispecific anti-IGF-1R and anti-ErbB3 antibody and one or more additional anti-cancer agents, wherein co-administration of the anti-cancer agent or agents has an additive or superadditive effect on suppressing tumor growth, as compared to administration of the bispecific anti-IGF-1R and anti-ErbB3 antibody alone or the one or more additional anti-cancer agents alone, wherein the effect on suppressing tumor growth is measured in a mouse xenograft model using BxPC-3, Caki-1, SK-ES-1, A549, NCI/ADR-RES, BT-474, DU145, or MCF7 cells.
  • kits comprising a therapeutically effective amount of a bispecific anti-IGF-1R and anti-ErbB3 antibody and a pharmaceutically-acceptable carrier.
  • the kits further comprise instructions to a practitioner, wherein the instructions comprise dosages and administration schedules for the bispecific anti-IGF-1R and anti-ErbB3 antibody.
  • the kit includes multiple packages each containing a single dose amount of the antibody.
  • the kit provides infusion devices for administration of the bispecific anti-IGF-1R and anti-ErbB3 antibody.
  • the kit further comprises an effective amount of at least one additional anti-cancer agent.
  • FIG. 1 is a graph demonstrating the inhibition of growth of Caki-1 renal cell carcinoma cancer cells in vivo by P4-G1-M1.3 (500 ⁇ g, 300 ⁇ g, or 100 ⁇ g) the mTOR inhibitor (mTORi) everolimus (30 mpk or 3 mpk), or the combination of everolimus (3 mpk) and P4-G1-M1.3 (500 ⁇ g).
  • the y-axis represents mean tumor volume in mm 3 and the x-axis represents time in days.
  • FIGS. 2 A-J are graphs demonstrating the level of IGF-1R and insulin receptor ( FIG. 2A ), EGFR and ErbB3 ( FIG. 2B ), ErbB2 ( FIG. 2C ), phospho-AKT (pAKT, Ser473 and Thr308) ( FIG. 2D ), phospho-Fox01 (Thr24)/Fox03a (Thr32) and phospho-PDK1 (pPDK1) ( FIG. 2E ), phospho-mTOR (p-mTOR) Ser2448 and Ser2481 ( FIG. 2F ), pS6 (Ser235/236 and Ser240/244)( FIG. 2G ), phospho-ERK (p-ERK) and survivin ( FIG.
  • FIGS. 3 A-D are graphs demonstrating the level of pAkt Ser473 ( FIG. 3A , B) and pERK ( FIG. 3C , D) in BxPC-3 cells ( FIG. 3A , C) wild-type for KRAS or KP4 cells ( FIG. 3B , D) mutant for KRAS.
  • Cells were treated with 500 nM P4-G1-M1.3, 250 nM GSK1120212 or the combination for 24 hours in 10% serum and ELISA assays were performed. The data was normalized to 10% serum without treatment.
  • FIG. 4 is a graph that demonstrates the inhibition of growth of DU145 prostate cancer cells in vivo by P4-G1-M1.3 alone (30 mpk, q3d), docetaxel alone (10 mpk q7d), or the combination of docetaxel and P4-G1-M1.3.
  • the y-axis represents mean tumor volume in mm 3 and the x-axis represents time in days.
  • FIGS. 5 A-D are graphs that demonstrate the level of ErbB3 ( FIG. 5A ), pErbB3 ( FIG. 5B ), pAkt Ser473 ( FIG. 5C ) and pERK1/2 ( FIG. 5D ) in HepG2 hepatocellular carcinoma cells.
  • Cells were treated with 500 nM P4-G1-M1.3, 5 ⁇ M sorafenib or the combination for either 2 hours or 6 hours and quantitative western blotting was performed.
  • FIG. 6 is a graph that represents the in vivo effects of P4-G1-M1.3 alone, docetaxel alone, or the combination of P4-G1-M1.3 and docetaxel on total IGF-1R in DU145 xenografts. Statistical significance across groups was determined using the student's T-test (*,p ⁇ 0.05 vs control.; #,p ⁇ 0.05 vs Docetaxel; a,p ⁇ 0.05 vs P4-G1-M1.3).
  • FIG. 7 is a graph that represents the in vivo effects of P4-G1-M1.3 alone, docetaxel alone, or the combination of P4-G1-M1.3 and docetaxel on total ErbB3 in DU145 xenografts. Statistical significance across groups was determined using the student's T-test (*, p ⁇ 0.05 vs control; #,p ⁇ 0.05 vs Docetaxel; a,p ⁇ 0.05 vs P4-G1-M1.3).
  • Methods of monotherapy, combination therapy, monotherapeutic compositions, and combination compositions for treating cancer in a patient are provided.
  • the cancer patient is treated with both a bispecific anti-IGF-1R and anti-ErbB3 antibody and one or more additional anti-cancer agents selected, e.g., from an mTOR inhibitor, a MEK inhibitor, a multikinase inhibitor, a B-Raf inhibitor, nanoliposomal irinotecan, a PI3K inhibitor, and an antimetabolite.
  • combination therapy with an effective amount of a first agent and an effective amount of a second agent provides a benefit that is greater than the benefit obtained in two matched comparisons: one in which the same effective amount of the first agent alone is separately administered as monotherapy to separate matched subjects and the other in which the same effective amount of the second agent alone is separately administered as monotherapy to separate matched subjects.
  • Such a greater benefit may be seen in patients treated with the combination therapy as an improved therapeutic outcome compared to either of the monotherapy comparators, or as a therapeutic outcome that is equal to or better than that of either of the monotherapy comparators and is associated in the combination therapy with a reduction of adverse events as compared to the adverse events seen with either of the monotherapy comparators.
  • An exemplary combinatorially enhanced outcome is one in which the greater benefit is a statistically significantly greater benefit with a p value of 0.05 or better, and each combinatorially enhanced outcome recited in the examples optionally corresponds to a statistically significantly greater benefit with a p value less than or equal to 0.05.
  • combination therapy includes simultaneous administration of at least two therapeutic agents to a patient or their sequential administration within a time period during which the first administered therapeutic agent is still present in the patient when the second administered therapeutic agent is administered.
  • the term “monotherapy” refers to administering a single drug to treat a disease or disorder in the absence of co-administration of any other therapeutic agent that is being administered to treat the same disease or disorder.
  • Additional anti-cancer agent is used herein to indicate any drug that is useful for the treatment of a malignant pancreatic tumor other than a drug that inhibits heregulin binding to ErbB2/ErbB3 heterodimer.
  • Dosage refers to parameters for administering a drug in defined quantities per unit time (e.g., per hour, per day, per week, per month, etc.) to a patient. Such parameters include, e.g., the size of each dose. Such parameters also include the configuration of each dose, which may be administered as one or more units, e.g., taken at a single administration, e.g., orally (e.g., as one, two, three or more pills, capsules, etc.) or injected (e.g., as a bolus). Dosage sizes may also relate to doses that are administered continuously (e.g., as an intravenous infusion over a period of minutes or hours). Such parameters further include frequency of administration of separate doses, which frequency may change over time.
  • Dose refers to an amount of a drug given in a single administration.
  • Effective amount refers to an amount (administered in one or more doses) of an antibody, protein or additional therapeutic agent, which amount is sufficient to provide effective treatment.
  • ErbB3 and “HERS” refer to ErbB3 protein, as described in U.S. Pat. No. 5,480,968.
  • the human ErbB3 protein sequence is shown in SEQ ID NO:4 of U.S. Pat. No. 5,480,968, wherein the first 19 amino acids (aas) correspond to the leader sequence that is cleaved from the mature protein.
  • ErbB3 is a member of the ErbB family of receptors, other members of which include ErbB1 (EGFR), ErbB2 (HER2/Neu) and ErbB4.
  • ErbB3 itself lacks tyrosine kinase activity, it can be phosphorylated upon dimerization with another ErbB family receptor, e.g., ErbB1, ErbB2 and ErbB4, which are receptor tyrosine kinases.
  • Ligands for the ErbB family include heregulin (HRG), betacellulin (BTC), epidermal growth factor (EGF), heparin-binding epidermal growth factor (HB-EGF), transforming growth factor alpha (TGF- ⁇ ), amphiregulin (AR), epigen (EPG) and epiregulin (EPR).
  • HRG heregulin
  • BTC betacellulin
  • EGF epidermal growth factor
  • HB-EGF heparin-binding epidermal growth factor
  • TGF- ⁇ transforming growth factor alpha
  • AR amphiregulin
  • EPG epigen
  • EPR epiregulin
  • IGF-1R refers to the receptor for insulin-like growth factor 1 (IGF-1, formerly known as somatomedin C). IGF-1R also binds to, and is activated by, insulin-like growth factor 2 (IGF-2). IGF1-R is a receptor tyrosine kinase, which upon activation by IGF-1 or IGF-2 is auto-phosphorylated.
  • IGF-1R insulin-like growth factor 1
  • IGF-2 insulin-like growth factor 2
  • IGF1-R is a receptor tyrosine kinase, which upon activation by IGF-1 or IGF-2 is auto-phosphorylated.
  • Genbank Accession No. NP — 000866 is assigned Gene ID: 3480.
  • Module refers to a structurally and/or functionally distinct part of a PBA, such a binding site (e.g., an scFv domain or a Fab domain) and the Ig constant domain. Modules provided herein can be rearranged (by recombining sequences encoding them, either by recombining nucleic acids or by complete or fractional de novo synthesis of new polynucleotides) in numerous combinations with other modules to produce a wide variety of PBAs, e.g., as disclosed herein.
  • an “SF” module refers to the binding site “SF,” i.e., comprising at least the CDRs of the SF VH and SF VL domains.
  • a “C8” module refers to the binding site “C8.”
  • PBA refers to a polyvalent bispecific antibody, an artificial hybrid protein comprising at least two different binding moieties or domains and thus at least two different binding sites (e.g., two different antibody binding sites), wherein one or more of the pluralities of the binding sites are covalently linked, e.g., via peptide bonds, to each other.
  • a preferred PBA described herein is an anti-IGF-1R+anti-ErbB3 PBA, which is a polyvalent bispecific antibody that comprises one or more first binding sites binding specifically to an IGF-1R protein, e.g., a human IGF-1R protein, and one or more second binding sites binding specifically to an ErbB3 protein, e.g., a human ErbB3 protein.
  • An anti-IGF-1R+anti-ErbB3 PBA is so named regardless of the relative orientations of the anti-IGF-1R and anti-ErbB3 binding sites in the molecule, whereas when the PBA name comprises two antigens separated by a slash (/) the antigen to the left of the slash is amino terminal to the antigen to the right of the slash.
  • a PBA may be a bivalent binding protein, a trivalent binding protein, a tetravalent binding protein or a binding protein with more than 4 binding sites.
  • An exemplary PBA is a tetravalent bispecific antibody, i.e., an antibody that has 4 binding sites, but binds to only two different antigens or epitopes.
  • Exemplary bispecific antibodies are tetravalent “anti-IGF-1R/anti-ErbB3” PBAs and “anti-ErbB3/anti-IGF-1R” PBAs.
  • the N-terminal binding sites of a tetravalent PBA are Fabs and the C-terminal binding sites are scFvs.
  • IGF-1R+ErbB3 PBAs comprising IgG1 constant regions each comprise two joined essentially identical subunits, each subunit comprising a heavy and a light chain that are disulfide bonded to each other, e.g., M7-G1-M78 (SEQ ID NO:146 and SEQ ID NO:147), P4-G1-M1.3 (SEQ ID NO:148 and SEQ ID NO:149), and P4-G1-C8 (SEQ ID NO:150 and SEQ ID NO:151), are exemplary embodiments of such IgG1-(scFv) 2 proteins.
  • the immunoglobulin constant regions are those of IgG2, the protein is referred to as an IgG2-(scFv) 2 .
  • IGF-1R+ErbB3 PBAs comprising IgG1 constant regions include, e.g., SF-G1-P1, SF-G1-M1.3, SF-G1-M27, SF-G1-P6, SF-G1-B69, P4-G1-C8, P4-G1-P1, P4-G1-M1.3, P4-G1-M27, P4-G1-P6, P4-G1-B69, M78-G1-C8, M78-G1-P1, M78-G1-M1.3, M78-G1-M27, M78-G1-P6, M78-G1-B69, M57-G1-C8, M57-G1-P1, M57-G1-M1.3, M57-G1-M27, M57-G1-P6, M57-G1-B69, P1-G1-P4, P1-G1-M57, P1-G1-M78, M27,
  • BPAs e.g., P4-G1-M1.3
  • additional anti-cancer agents e.g., an mTOR inhibitor, a MEK inhibitor, a multikinase inhibitor, a B-Raf inhibitor, an anti-endocrine therapy, antihormonal therapy, irinotecan or nanoliposomal irinotecan, a PI3K inhibitor, or an antimetabolite
  • a cancer e.g., pancreatic, ovarian, lung, colon, head and neck, and esophageal cancers.
  • Additional anti-cancer agents suitable for combination with anti-IGF-1R+anti-ErbB3 antibodies may include but are not limited to pyrimidine antimetabolites (e.g., the nucleoside metabolic inhibitor gemcitabine, cytarabine, or the pyrimidine analog 5-fluorouracil), mTOR inhibitors (e.g., everolimus, temsirolimus, sirolimus, or ridaforolimus), pan-mTOR inhibitors (e.g., INK128, CC223, OS1207, AZD8055, AZD2014, or Palomid529), phosphoinositide-3-kinase (PI3K) inhibitors (e.g., perifosine (KRX-0401), SF1126, CAL101, BKM120, BKM120, XL147, and PX-866), MEK inhibitors (e.g., GSK1120212, BAY 86-9766 or AZD624), taxanes (e.
  • one or more of the following therapeutic agents is co-administered to the patient with an anti-IGF-1R+anti-ErbB3 antibody.
  • Gemcitabine (Gemzar®) is indicated as first line therapy for pancreatic adenocarcinoma and is also used in various combinations to treat ovarian, breast and non-small-cell lung cancers.
  • Temsirolimus (Torisel®) is an mTOR inhibitor that is administered parenterally, typically by i.v. infusion and is used to treat advanced renal cell carcinoma.
  • Everolimus (Afinitor®), a 40-O-(2-hydroxyethyl) derivative of sirolimus, is an mTOR inhibitor that is administered orally and is used to treat progressive neuroendocrine tumors of pancreatic origin (PNET) in patients with unresectable, locally advanced or metastatic disease.
  • PNET pancreatic origin
  • 5-Fluorouracil (5-FU Adrucil®, Carac®, Efudix®, Efudex® and Fluoroplex®) is a pyrimidine analog that works through irreversible inhibition of thymidylate synthase.
  • Capecitabine (Xeloda®) is an orally administered systemic prodrug of 5′-deoxy-5-fluorouridine (5′-DFUR) which is converted to 5-fluorouracil.
  • Docetaxel is an anti-mitotic chemotherapy used for the treatment of breast, advanced non-small cell lung, metastatic androgen-independent prostate, advanced gastric and locally advanced head and neck cancers.
  • Paclitaxel (Taxol®) is an anti-mitotic chemotherapy used for the treatment of lung, ovarian, breast and head and neck cancers.
  • Sorafenib (Nexavar®) is a small molecule inhibitor of multiple tyrosine kinases (including VEGFR and PDGFR) and Raf kinases (an exemplary “multikinase inhibitor”) used for treatment of advanced renal cell carcinoma (RCC) and advanced primary liver cnacer (hepatocellular carcinoma, HCC).
  • RRC renal cell carcinoma
  • HCC hepatocellular carcinoma
  • Trametinib (GSK-1120212) is a small molecule inhibitor of the MEK protein currently in clinical trials for the treatment of several cancers including pancreatic, melanoma, breast and non-small cell lung.
  • Vemurafenib (Zelboraf®) is a small molecule inhibitor of B-Raf in patients whose cancer cells harbor a V600E B-Raf mutation. Vemurafenib is currently approved for treatment of late-stage, unresectable, and metastatic melanoma.
  • Nanoliposomal irinotecan (e.g., MM-398) is a stable nanoliposomal formulation of irinotecan.
  • MM-398 is described, e.g., in U.S. Pat. No. 8,147,867.
  • MM-398 may be administered, for example, on day 1 of the cycle at a dose of 120 mg/m2, except if the patient is homozygous for allele UGT1A1*, wherein nanoliposomal irinotecan is administered on day 1 of cycle 1 at a dose of 80 mg/m 2 .
  • the required amount of MM-398 may be diluted, e.g., in 500 mL of 5% dextrose injection USP and infused over a 90 minute period.
  • co-administration of an anti-IGF-1R+anti-ErbB3 antibody with one or more additional therapeutic agents provides improved efficacy compared to treatment with the antibody alone or with the one or more additional therapeutic agents in the absence of antibody therapy.
  • additional therapeutic agents e.g., everolimus, temsirolimus, sirolimus, XL147, gemcitabine, 5-fluorouracil, cytarabine
  • a combination of an anti-IGF-1R+anti-ErbB3 antibody with one or more additional therapeutic agents exhibits therapeutic synergy.
  • “Therapeutic synergy” refers to a phenomenon where treatment of patients with a combination of therapeutic agents manifests a therapeutically superior outcome to the outcome achieved by each individual constituent of the combination used at its optimum dose (T. H. Corbett et al., 1982, Cancer Treatment Reports, 66, 1187).
  • a therapeutically superior outcome is one in which the patients either a) exhibit fewer incidences of adverse events while receiving a therapeutic benefit that is equal to or greater than that where individual constituents of the combination are each administered as monotherapy at the same dose as in the combination, or b) do not exhibit dose-limiting toxicities while receiving a therapeutic benefit that is greater than that of treatment with each individual constituent of the combination when each constituent is administered in at the same doses in the combination(s) as is administered as individual components.
  • a combination, used at its maximum tolerated dose, in which each of the constituents will be present at a dose generally not exceeding its individual maximum tolerated dose manifests therapeutic synergy when decrease in tumor growth achieved by administration of the combination is greater than the value of the decrease in tumor growth of the best constituent when the constituent is administered alone.
  • the components of such combinations have an additive or superadditive effect on suppressing tumor growth, as compared to monotherapy with the PBA or treatment with the chemotherapeutic(s) in the absence of antibody therapy.
  • additive is meant a result that is greater in extent (e.g., in the degree of reduction of tumor mitotic index or of tumor growth or in the degree of tumor shrinkage or the frequency and/or duration of symptom-free or symptom-reduced periods) than the best separate result achieved by monotherapy with each individual component, while “superadditive” is used to indicate a result that exceeds in extent the sum of such separate results.
  • the additive effect is measured as slowing or stopping of tumor growth.
  • the additive effect can also be measured as, e.g., reduction in size of a pancreatic tumor, reduction of tumor mitotic index, reduction in number of metastatic lesions over time, increase in overall response rate, or increase in median or overall survival.
  • T C represents the delay in growth of the cells, which is the average time, in days, for the tumors of the treated group (T) and the tumors of the control group (C) to have reached a predetermined value (1 g, or 10 mL, for example), and Td represents the time, in days necessary for the volume of the tumor to double in the control animals.
  • a combination, used at its own maximum tolerated dose, in which each of the constituents is present at a dose generally less than or equal to its maximum tolerated dose exhibits therapeutic synergy when the log 10 cell kill is greater than the value of the log 10 cell kill of the best constituent when it is administered alone.
  • the log 10 cell kill of the combination exceeds the value of the log 10 cell kill of the best constituent of the combination by at least 0.1 log cell kill, at least 0.5 log cell kill, or at least 1.0 log cell kill.
  • kits that include a pharmaceutical composition containing a bispecific anti-IGF-1R and anti-ErbB3 antibody, including a pharmaceutically-acceptable carrier, in a therapeutically effective amount adapted for use in the preceding methods.
  • the kits include instructions to allow a practitioner (e.g., a physician, nurse, or patient) to administer the composition contained therein to treat an ErbB2 expressing cancer.
  • kits include multiple packages of the single-dose pharmaceutical composition(s) containing an effective amount of a bispecific anti-IGF-1R and anti-ErbB3 antibody for a single administration in accordance with the methods provided above.
  • instruments or devices necessary for administering the pharmaceutical composition(s) may be included in the kits.
  • a kit may provide one or more pre-filled syringes containing an amount of a bispecific anti-IGF-1R and anti-ErbB3 antibody that is about 100 times the dose in mg/kg indicated for administration in the above methods.
  • kits may also include additional components such as instructions or administration schedules for a patient suffering from a disease or condition (e.g., a cancer, autoimmune disease, or cardiovascular disease) to use the pharmaceutical composition(s) containing a bispecific anti-IGF-1R and anti-ErbB3 antibody, or any binding, diagnostic, and/or therapeutic agent conjugated thereto.
  • a disease or condition e.g., a cancer, autoimmune disease, or cardiovascular disease
  • BxPC-3 mouse xenograft models are established using 5 ⁇ 10 6 BxPC-3 cells that are resuspended 1:1 with PBS:Growth factor-reduced Matrigel® and injected subcutaneously into Nu/Nu mice. Tumors are allowed to develop for 8 days. Antibodies are injected intraperitoneally every 3 days (q3d) for 2 rounds of dosing.
  • mice For the BxPC-3 PD study, 4 treatment groups are established, each containing 4 mice. These included control, P4-G1-M1.3 (q3d, 600 ⁇ g), gemcitabine (q3d, 150 mg/kg) and P4-G1-M1.3+gemcitabine (combined individual doses). Tumors are excised on either day 19 or day 28, resulting in a total of 8 groups.
  • Tumors are initially weighed and pulverized in a CryoPrep® tissue pulverizer (Covaris). Tissue Extraction Reagent 1 (TER1, Life TechnologiesTM) containing protease and phosphatase inhibitors was added to the tumor at a ratio of 1 ml TER1 per 100 mg of tissue. Samples are incubated on ice for 30 minutes to solubilize tissue and put through a QIAshredderTM column (Qiagen) according to the manufacturer's protocol. A BCA assay (Pierce) is performed to determine protein concentration according to the manufacturer's protocol.
  • TER1 Tissue Extraction Reagent 1
  • Qiagen QIAshredderTM column
  • Buffer containing ⁇ -Mercaptoethanol ( ⁇ -ME) is added and lysates are boiled for 5 minutes at 95° C. Approximately 40 ⁇ g of protein and two ladders (Invitrogen) are run on each well of an 18-well gel (BioRad). Gels are run at 150 volts constant for approximately 90 minutes and transferred to nitrocellulose membranes using the 8 minute transfer program of the iBlot® (Invitrogen) transfer system. Membranes are blocked in Odyssey® Blocking Buffer (Licor@ Biosciences) for 1 hour at room temperature, and then incubated with primary antibodies overnight at 4° C. in 5% BSA in TBS-T. All antibodies are purchased from Cell Signaling and used at the recommended dilution.
  • ⁇ -ME ⁇ -Mercaptoethanol
  • membranes are washed 3 ⁇ 5 minutes each with TBS-T and then incubated with anti-Rabbit IgG-DyLight® 800 (Cell Signaling) or anti-Rabbit IRDye® 800 (Licor@ Biosciences) at 1:10,000-15,000 in 5% milk in TBS-T for 1 hour at room temperature.
  • Membranes were then washed 3 ⁇ 5 minutes each with TBS-T and scanned using the Licor® Odyssey® system (Licor@ Biosciences). Intensities are quantified using Image Studio 2.0 and normalized to ⁇ -Actin levels.
  • Patients with renal cell carcinoma are treated by administration of monotherapy with either an effective amount of mTOR inhibitor everolimus (Afinitor®) or an effective amount of P4-G1-M1.3, or with combination therapy comprising or consisting of administration of both the effective amount of everolimus and the effective amount of P4-G1-M1.3.
  • an effective amount of mTOR inhibitor everolimus Afinitor®
  • an effective amount of P4-G1-M1.3 or with combination therapy comprising or consisting of administration of both the effective amount of everolimus and the effective amount of P4-G1-M1.3.
  • P4-G1-M1.3 is formulated in 20 mM histidine, 100 mM arginine-HCl, 3% sucrose, at pH 5.5 supplemented with 0.002-0.02% of Tween® 80 at concentration range 5-15 mg/mL.
  • P4-G1-M1.3 is administered to patients at 6 mg/kg, 12 mg/kg, 20 mg/kg, 30 mg/kg, 40 mg/kg, 50 mg/kg or 60 mg/kg q7d, q14d, q21d, or q28d with a loading dose of 12 mg/kg, 20 mg/kg, 40 mg/kg, 40 mg/kg or 60 mg/kg
  • Everolimus is administered to patients at 2.5 mg, 5 mg, or 10 mg orally once a day or once every other day.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • Patients with gastrointestinal neuroendocrine tumors are treated by administration of either monotherapy with an effective amount of everolimus or an effective amount of P4-G1-M1.3, or with combination therapy comprising or consisting of administration of both the effective amount of everolimus and the effective amount of P4-G1-M1.3.
  • P4-G1-M1.3 and everolimus are prepared and dosed as described in Example 1. The combination therapy will provide a combinatorially enhanced outcome.
  • Example 3 The advantages of combination therapy per Example 3 are demonstrated in a preclinical model carried out using the methods of Example 2 adapted for the substitution of human pancreatic adenocarcinoma BXPC-3 cells for the Caki-1 cells of Example 2. The results will demonstrate that P4-G1-M1.3 suppresses tumor growth of BXPC-3 cells in vivo and potentiates responses to everolimus.
  • NSCLC non-small cell lung cancer
  • Example 5 The advantages of combination therapy per Example 5 are demonstrated in a preclinical model carried out using the methods of Example 2 adapted for the substitution of human NSCLC A549 cells for the Caki-1 cells of Example 2. The results will demonstrate that P4-G1-M1.3 suppresses tumor growth of A549 cells in vivo and potentiates responses to everolimus.
  • Patients with gastrointestinal neuroendocrine tumors are treated by administration of either monotherapy with an effective amount of mTOR inhibitor temsirolimus (Torisel®) or an effective amount of P4-G1-M1.3, or with combination therapy comprising or consisting of administration of both the effective amount of temsirolimus and the effective amount of P4-G1-M1.3.
  • P4-G1-M1.3 is prepared and dosed as described in Example 1. Temsirolimus is dosed at 2.5 mg, 7.5 mg, 15 mg, or 25 mg (25 mg is the manufacturer's recommended dose) infused over a 30-60 minute period once a week.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • Example 7 The advantages of combination therapy per Example 7 are demonstrated in a preclinical model carried out using the methods of Example 2 adapted for the substitution of human pancreatic adenocarcinoma BXPC-3 cells for the Caki-1 cells of Example 2. The results will show that P4-G1-M1.3 suppresses tumor growth of BXPC-3 cells in vivo and potentiates the response to everolimus.
  • Example 9 The advantages of combination therapy per Example 9 are demonstrated in a preclinical model carried out using the methods of Example 2 adapted for the substitution of SK-ES-1 human Ewing sarcoma cells for the Caki-1 cells of Example 2.
  • the results will show that P4-G1-M1.3 suppresses tumor growth of SK-ES-1 cells in vivo and potentiates responses to everolimus.
  • Patients with gastrointestinal neuroendocrine tumors are treated by administration of either monotherapy with an effective amount of mTOR inhibitor sirolimus (Rapamune®) or an effective amount of P4-G1-M1.3, or with combination therapy comprising or consisting of administration of both the effective amount of sirolimus and the effective amount of P4-G1-M1.3.
  • P4-G1-M1.3 is prepared and dosed as described in Example 1. Patients are dosed with sirolimus at 0.2 mg, 0.5 mg, 2 mg, 5 mg, 10 mg, 15 mg, or 20 mg orally once a day with a loading dose of 0.6 mg, 1.5 mg, 6 mg, 15 mg, or 30 mg (3 ⁇ the maintenance dose.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • Patients with estrogen receptor positive or progesterone receptor positive or triple negative breast cancers that are locally advanced or metastatic, or with any of the tumor types listed in Examples 5-10, are treated with a combination of an effective amount of i) any of the mTOR inhibitors of the preceding examples (at doses as described therein), or with a pan-mTOR inhibitor (INK128, CC223, OSI207, AZD8055, AZD2014, or Palomid529) and ii) an effective amount of P4-G1-M1.3.
  • Patients are dosed with P4-G1-M1.3 as described in Example 1.
  • the dose of pan-mTOR inhibitor is the dose used in phase I or, preferably phase II or III clinical trials.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • Postmenopausal women with estrogen receptor-positive locally advanced or metastatic breast cancer are treated with a combination of an effective amount of PI3K inhibitor (e.g., XL147 or BKM120) and an effective amount of P4-G1-M1.3.
  • Patients are dosed with P4-G1-M1.3 as described in Example 1.
  • XL147 is dosed at 25, 50, 100, or 200 mg orally once a day for 21 consecutive days.
  • BKM120 is dosed at 12.5, 25, 50, 100, or 20 mg orally once a day for 28 consecutive days.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • Women with estrogen or progesterone receptor positive locally advanced or metastatic breast cancer are treated with a combination therapy comprising or consisting of administration of an effective amount of an antihormonal therapy (such as tamoxifen, exemestane, letrozole or fulvestrant) and administration of an effective amount of P4-G1-M1.3.
  • P4-G1-M1.3 is formulated and administered as described above.
  • Antihormonal therapy is administered in accordance with manufacturer's directions.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • Women with estrogen or progesterone receptor positive locally advanced or metastatic breast cancer are treated with a combination of an effective amount of PI3K/mTOR dual inhibitor NVP-BEZ235 and an effective amount of P4-G1-M1.3. Patients are dosed with P4-G1-M1.3 as described in Example 1.
  • NVP-BEZ235 is given orally twice daily at doses of 400 mg, 600 mg, or 800 mg. The combination therapy will provide a combinatorially enhanced outcome.
  • the preclinical models to demonstrate the working of this Example is adapted from Examples 2, 4, 6, 8, 10, 12, and 15 using the ZR-75-1 human breast cancer cells and Caki-1 cells.
  • the combination therapies of Examples 1, 11, 13, 14 and 17 are expanded to include patients in which the tumor type can be pancreatic cancer, Ewing's sarcoma family of tumors, NSCLC, renal cell carcinoma (second line in renal carcinoma patients refractory to sunitinib (Sutent®)), or estrogen or progesterone receptor positive locally advanced or metastatic breast cancer.
  • the tumor type can be pancreatic cancer, Ewing's sarcoma family of tumors, NSCLC, renal cell carcinoma (second line in renal carcinoma patients refractory to sunitinib (Sutent®)), or estrogen or progesterone receptor positive locally advanced or metastatic breast cancer.
  • Patients with pancreatic carcinoma are treated with a combination of an effective amount of gemcitabine, cytarabine, capecitabine, or 5-fluorouracil (5-FU) and an effective amount of P4-G1-M1.3.
  • Patients are dosed with P4-G1-M1.3 as described in Example 1.
  • Patients are dosed with the manufacturer's recommended dose of gemcitabine, capecitabine or 5-FU.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • Preclinical xenograft data to support this Example were obtained using the pancreatic carcinoma cancer model BxPC-3.
  • BxPC-3 xenografts mice with control tumors were compared to those treated with monotherapy with P4-G1-M1.3, monotherapy with gemcitabine or combination therapy with P4-G1-M1.3 and gemcitabine.
  • P4-G1-M1.3 downregulates receptor complexes and inhibits PI3K/AKT/mTOR signaling in BxPC-3 PD study tumors. Results appear in the figures as follows: downregulation of IGF-1R and Insulin Receptor ( FIG. 2A ), EGFR and ErbB3 ( FIG. 2B ), ErbB2 ( FIG.
  • P4-G1-M1.3 inhibits ERK phosphorylation and potentiates apoptosis-inducing activity of gemcitabine ( FIG. 2H )
  • tumor types that can be beneficially treated with effective amounts of bispecific anti-IGF-1R and anti-ErbB3 antibodies disclosed herein in combinations administered in accordance with this disclosure include thyroid carcinoma, head and neck squamous cell carcinoma, breast carcinoma, lung cancer (e.g., small-cell lung carcinoma, non-small-cell lung carcinoma), gastric carcinoma, gastrointestinal stromal tumors, ovarian carcinoma, bile duct carcinoma, endometrial carcinoma, prostate carcinoma, renal cell carcinoma, anaplastic large-cell lymphoma, leukemia (e.g., acute myeloid leukemia, T-cell leukemia, chronic lymphocytic leukemia), multiple myeloma, malignant mesothelioma, malignant melanoma, colon cancer, sarcoma.
  • leukemia e.g., acute myeloid leukemia, T-cell leukemia, chronic lymphocytic leukemia
  • multiple myeloma malignant mesothelioma, mal
  • Patients with pancreatic cancer are treated with a combination of an effective amount of a MEK inhibitor (e.g., GSK1120212, BAY 86-9766 or AZD6244) and an effective amount of P4-G1-M1.3.
  • a MEK inhibitor e.g., GSK1120212, BAY 86-9766 or AZD6244
  • P4-G1-M1.3 an effective amount of a MEK inhibitor
  • Patients are dosed with P4-G1-M1.3 as described in Example 1.
  • the clinical dose of MEK inhibitor is the dose used for that inhibitor in phase II or phase III clinical trials.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • treatment of cancer cells in vitro with GSK1120212 and P4-G1-M1.3 results in inhibition of signaling in both a wild-type and KRAS mutant background.
  • the preclinical models to support this example are performed using BxPC-3 (KRAS wild-type) and KP4 (KRAS mutant) cell lines.
  • Women with locally advanced or metastatic breast cancer are treated with a combination of an effective amount of docetaxel (Taxotere®) and an effective amount of P4-G1-M1.3.
  • Patients are dosed with P4-G1-M1.3 as described in Example 1.
  • Patients are dosed with docetaxel at 25, 50, 75 or 100 mg/m 2 i.v. once every 3 weeks or per standard clinical practice.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • FIG. 4 the combination of docetaxel and P4-G1-M1.3 results in inhibition of growth of DU145 prostate cancer cells in vivo.
  • FIGS. 6 and 7 demonstrate the in vivo effects of P4-G1-M1.3, Docetaxel, or the combination on total IGF-1R ( FIG. 6 ) and total ErbB3 ( FIG. 7 ) in DU145 xenografts. Statistical significance across groups was determined using the student's T-test (*,p ⁇ 0.05 vs control.; #,p ⁇ 0.05 vs docetaxel; a,p ⁇ 0.05 vs P4-G1-M1.3).
  • Patients with metastatic breast cancer are treated with a combination of an effective amount of paclitaxel (Taxol®) or of eribulin and an effective amount of P4-G1-M1.3. Patients are dosed with P4-G1-M1 as described in Example 1. Patients are dosed per standard clinical practice for paclitaxel or eribulin. The combination therapy will provide a combinatorially enhanced outcome.
  • Example 24 The treatments of Example 24 are repeated in patients where the tumor type is squamous cell carcinoma of the lung, prostate cancer or ovarian cancer. The results will be the same as obtained in Example 24.
  • HCC hepatocellular carcinoma
  • Patients with hepatocellular carcinoma are treated with P4-G1-M1.3 monotherapy. Patients are dosed with P4-G1-M1.3 as described in Example 1 (second line in patients refractory to sorafenib). Patients will obtain a statistically significant improvement in HCC symptoms (e.g., time to progression or progression-free survival at pre-defined intervals) compared to untreated historical controls or to best supportive care. Preclinical data to support this example may be obtained using HepG2 cells in vitro and in vivo.
  • HCC hepatocellular carcinoma
  • patients with hepatocellular carcinoma are treated with a combination of an effective amount of sorafenib and an effective amount of P4-G1-M1.3.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • Preclinical data to support this example may be obtained using HepG2 cells in vitro and in vivo. As shown in FIG.
  • treatment of HepG2 hepatocellular carcinoma cells with the combination of sorafenib and P4-G1-M1.3 results in downregulation of ErbB3 and inhibits downstream signaling when compared to treatments with sorafenib alone or with P4-G1-M1.3 alone.
  • Patients with melanoma are treated by administration of either monotherapy with an effective amount of vemurafenib (Zelboraf®) or an effective amount of P4-G1-M1.3, or with combination therapy comprising or consisting of administration of both the effective amount of vemurafenib or dabrafenib and the effective amount of P4-G1-M1.3.
  • P4-G1-M1.3 is formulated and administered as described above in Example 1.
  • Vemurafenib is given orally at 960 mg 1-2 times daily.
  • Dabrafenib is administered as administered in dabrafenib phase III clinical trials.
  • the combination therapy will provide a combinatorially enhanced outcome.
  • irinotecan (Camptosar®) or nanoliposomal irinotecan and an effective amount of P4-G1-M1.3.
  • P4-G1-M1.3 is formulated and administered as described above (e.g., Example 1).
  • Nanoliposomal irinotecan is given i.v. at 80 mg/m 2 or 120 mg/m 2 q3w. Camptosar® is administered per manufacturer's directions.
  • the combination therapy will provide a combinatorially enhanced outcome.
US14/388,330 2012-04-02 2013-04-02 Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies Abandoned US20150231219A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/388,330 US20150231219A1 (en) 2012-04-02 2013-04-02 Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261619258P 2012-04-02 2012-04-02
US201261723582P 2012-11-07 2012-11-07
PCT/US2013/035013 WO2013152034A1 (fr) 2012-04-02 2013-04-02 Dosage et administration d'anticorps anti-igf-1r et anti-erbb3 monospécifiques et bispécifiques
US14/388,330 US20150231219A1 (en) 2012-04-02 2013-04-02 Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/035013 A-371-Of-International WO2013152034A1 (fr) 2012-04-02 2013-04-02 Dosage et administration d'anticorps anti-igf-1r et anti-erbb3 monospécifiques et bispécifiques

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/597,781 Continuation US20180036395A1 (en) 2012-04-02 2017-05-17 Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies

Publications (1)

Publication Number Publication Date
US20150231219A1 true US20150231219A1 (en) 2015-08-20

Family

ID=48142962

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/388,330 Abandoned US20150231219A1 (en) 2012-04-02 2013-04-02 Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies
US15/597,781 Abandoned US20180036395A1 (en) 2012-04-02 2017-05-17 Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/597,781 Abandoned US20180036395A1 (en) 2012-04-02 2017-05-17 Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies

Country Status (13)

Country Link
US (2) US20150231219A1 (fr)
EP (1) EP2833915A1 (fr)
JP (1) JP2015514113A (fr)
KR (1) KR20140148412A (fr)
CN (1) CN104684579A (fr)
AU (1) AU2013243584A1 (fr)
BR (1) BR112014024494A2 (fr)
CA (1) CA2868516A1 (fr)
HK (1) HK1207000A1 (fr)
IL (1) IL234866A0 (fr)
IN (1) IN2014DN09098A (fr)
MX (1) MX2014011925A (fr)
WO (1) WO2013152034A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016196377A1 (fr) 2015-05-29 2016-12-08 Merrimack Pharmaceuticals, Inc. Polythérapies contre le cancer
US9527914B2 (en) * 2011-04-19 2016-12-27 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-IGF-1R and anti-ErbB3 antibodies
WO2017181099A1 (fr) * 2016-04-15 2017-10-19 Merrimack Pharmaceuticals, Inc. Dosage et administration d'anticorps bispécifiques anti-igf-1r et anti-erbb3, leurs utilisations et méthodes de traitement les mettant en oeuvre
US10322192B2 (en) 2016-03-02 2019-06-18 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
WO2022183185A1 (fr) * 2021-02-23 2022-09-01 Pandion Operations, Inc. Anticorps anti-pd-1, polypeptides et leurs utilisations

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7960139B2 (en) 2007-03-23 2011-06-14 Academia Sinica Alkynyl sugar analogs for the labeling and visualization of glycoconjugates in cells
JP5986745B2 (ja) 2008-07-15 2016-09-06 アカデミア シニカAcademia Sinica Ptfe様のアルミニウム・コート・ガラススライド上のグリカンアレイおよび関連する方法
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
US11377485B2 (en) 2009-12-02 2022-07-05 Academia Sinica Methods for modifying human antibodies by glycan engineering
DK2544680T3 (en) 2010-03-11 2015-04-27 Merrimack Pharmaceuticals Inc USE OF ErbB3 INHIBITORS IN TREATMENT OF TRIPLE-NEGATIVE BREAST CANCER
WO2011130332A1 (fr) 2010-04-12 2011-10-20 Academia Sinica Puces au glycane pour la recherche par criblage haut débit de virus
US10130714B2 (en) 2012-04-14 2018-11-20 Academia Sinica Enhanced anti-influenza agents conjugated with anti-inflammatory activity
AU2013306098A1 (en) 2012-08-18 2015-02-12 Academia Sinica Cell-permeable probes for identification and imaging of sialidases
US10086054B2 (en) 2013-06-26 2018-10-02 Academia Sinica RM2 antigens and use thereof
EP3013347B1 (fr) 2013-06-27 2019-12-11 Academia Sinica Conjugués de glycane et leur utilisation
CN105682666B (zh) 2013-09-06 2021-06-01 中央研究院 使用醣脂激活人类iNKT细胞
EP3082859A4 (fr) * 2013-12-19 2017-07-19 Medlmmune, LLC Compositions et méthodes pour traiter un sarcome
WO2015100459A2 (fr) 2013-12-27 2015-07-02 Merrimack Pharmaceuticals, Inc. Profils de biomarqueur pour prédire les résultats d'une thérapie cancéreuse utilisant des inhibiteurs d'erbb3 et/ou des chimiothérapies
US9982041B2 (en) 2014-01-16 2018-05-29 Academia Sinica Compositions and methods for treatment and detection of cancers
US10150818B2 (en) 2014-01-16 2018-12-11 Academia Sinica Compositions and methods for treatment and detection of cancers
WO2015130554A2 (fr) * 2014-02-20 2015-09-03 Merrimack Pharmaceuticals, Inc. Dosage et administration d'anticorps bispecifiques anti-igf-1r et anti-erbb3, leurs utilisations et procedes de traitement les mettant en oeuvre
WO2015134399A1 (fr) * 2014-03-03 2015-09-11 Eisai R&D Management Co., Ltd. Utilisation d'éribuline et d'inhibiteurs de mtor en tant que polythérapie pour le traitement du cancer
TWI687428B (zh) 2014-03-27 2020-03-11 中央研究院 反應性標記化合物及其用途
EP3149161B1 (fr) 2014-05-27 2021-07-28 Academia Sinica Fucosidase issue de bacteroïdes et ses procédés d'utilisation
CN106573971A (zh) 2014-05-27 2017-04-19 中央研究院 抗cd20醣抗体及其用途
AU2015267045B2 (en) 2014-05-27 2021-02-25 Academia Sinica Anti-HER2 glycoantibodies and uses thereof
US10118969B2 (en) 2014-05-27 2018-11-06 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
TWI732738B (zh) 2014-05-28 2021-07-11 中央研究院 抗TNF-α醣抗體及其用途
WO2016040369A2 (fr) 2014-09-08 2016-03-17 Academia Sinica Activation des cellules inkt humaines par des glycolipides
US10495645B2 (en) 2015-01-16 2019-12-03 Academia Sinica Cancer markers and methods of use thereof
US9975965B2 (en) 2015-01-16 2018-05-22 Academia Sinica Compositions and methods for treatment and detection of cancers
EP3248005B1 (fr) 2015-01-24 2020-12-09 Academia Sinica Nouveaux composés conjugués de glycane et leurs méthodes d'utilisation
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
US20170233491A1 (en) 2016-01-19 2017-08-17 Merrimack Pharmaceuticals, Inc. Dosage and administration of combination therapies comprising istiratumab, uses and methods of treatment
TW201808978A (zh) 2016-03-08 2018-03-16 中央研究院 N-聚醣及其陣列之模組化合成方法
JP7213549B2 (ja) 2016-08-22 2023-01-27 シーエイチオー ファーマ インコーポレイテッド 抗体、結合性断片、および使用の方法
MX2019008458A (es) 2017-01-17 2019-12-02 Heparegenix Gmbh Inhibidores de proteina cinasa para promover la regeneracion hepatica o reducir o prevenir la muerte de hepatocitos.

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120244163A1 (en) * 2009-10-14 2012-09-27 Dyax Corp. Bispecific binding agents targeting igf-1r and erbb3 signalling and uses thereof
US20120269812A1 (en) * 2011-04-19 2012-10-25 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-igf-1r and anti-erbb3 antibodies

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5183884A (en) 1989-12-01 1993-02-02 United States Of America Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
KR101462825B1 (ko) 2004-05-03 2014-11-21 헤르메스 바이오사이언스, 인코포레이티드 약물 전달에 유용한 리포좀

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120244163A1 (en) * 2009-10-14 2012-09-27 Dyax Corp. Bispecific binding agents targeting igf-1r and erbb3 signalling and uses thereof
US20120269812A1 (en) * 2011-04-19 2012-10-25 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-igf-1r and anti-erbb3 antibodies

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9527914B2 (en) * 2011-04-19 2016-12-27 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-IGF-1R and anti-ErbB3 antibodies
US9556274B2 (en) 2011-04-19 2017-01-31 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-IGF-1R and anti-ERBB3 antibodies
US9938346B2 (en) 2011-04-19 2018-04-10 Merrimack Pharmaceuticals, Inc. Monospecific and bispecific anti-IGF-1R and anti-ErbB3 antibodies
WO2016196377A1 (fr) 2015-05-29 2016-12-08 Merrimack Pharmaceuticals, Inc. Polythérapies contre le cancer
US10322192B2 (en) 2016-03-02 2019-06-18 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US10548986B2 (en) 2016-03-02 2020-02-04 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
WO2017181099A1 (fr) * 2016-04-15 2017-10-19 Merrimack Pharmaceuticals, Inc. Dosage et administration d'anticorps bispécifiques anti-igf-1r et anti-erbb3, leurs utilisations et méthodes de traitement les mettant en oeuvre
WO2022183185A1 (fr) * 2021-02-23 2022-09-01 Pandion Operations, Inc. Anticorps anti-pd-1, polypeptides et leurs utilisations

Also Published As

Publication number Publication date
IN2014DN09098A (fr) 2015-05-22
HK1207000A1 (en) 2016-01-22
US20180036395A1 (en) 2018-02-08
CA2868516A1 (fr) 2013-10-10
EP2833915A1 (fr) 2015-02-11
WO2013152034A1 (fr) 2013-10-10
AU2013243584A1 (en) 2014-10-09
KR20140148412A (ko) 2014-12-31
BR112014024494A2 (pt) 2017-08-08
JP2015514113A (ja) 2015-05-18
MX2014011925A (es) 2015-05-11
CN104684579A (zh) 2015-06-03
IL234866A0 (en) 2014-12-31

Similar Documents

Publication Publication Date Title
US20180036395A1 (en) Dosage and administration of monospecific and bispecific anti-igr-1r and anti-erbb3 antibodies
Capelan et al. Pertuzumab: new hope for patients with HER2-positive breast cancer
US20190119401A1 (en) Use of erbb3 inhibitors in the treatment of triple negative and basal-like breast cancers
JP6282296B2 (ja) Egfrを標的とする癌の組み合わせ治療方法
TWI780994B (zh) 用於治療非小細胞肺癌之抗b7-h1及抗ctla-4抗體
KR20140023921A (ko) ErbB 경로 저해제들에 대한 저항성을 극복하는 방법
L Hixon et al. Development of inhibitors of the IGF-IR/PI3K/Akt/mTOR pathway
US20170096492A1 (en) DOSAGE AND ADMINISTRATION OF ANTI-IGF-1R, ANTI-ErbB3 BISPECIFIC ANTIBODIES, USES THEREOF AND METHODS OF TREATMENT THEREWITH
WO2013138371A1 (fr) Procédés de traitement du cancer du pancréas à l'aide de thérapies combinées comprenant un anticorps anti-erbb3
Specenier et al. Biologic therapy in head and neck cancer: a road with hurdles
WO2014036520A1 (fr) Polythérapies comprenant des agents anti-erbb3
JP2020506945A (ja) がんの治療のための方法、組成物及びキット
Smith et al. Pertuzumab for the treatment of patients with previously untreated HER2-positive metastatic breast cancer
WO2019070497A1 (fr) Polythérapie contre le cancer
JP2020510058A (ja) トリフルリジン/チピラシル塩酸塩、抗腫瘍白金錯体、及び免疫チェックポイント調節剤の新規組み合わせ物
Patel EGFR signaling and its inhibition by EGFR inhibitors in NSCLC
WO2022107027A1 (fr) Administration d'un agoniste de sting, d'inhibiteurs de point de contrôle et de rayonnement

Legal Events

Date Code Title Description
AS Assignment

Owner name: MERRIMACK PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LUGOVSKOY, ALEXEY ALEXANDROVICH;BAUM, JASON;ADAMS, SHARLENE;AND OTHERS;REEL/FRAME:033786/0513

Effective date: 20140912

AS Assignment

Owner name: MERRIMACK PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LUGOVSKOY, ALEXEY ALEXANDROVICH;BAUM, JASON;ADAMS, SHARLENE;REEL/FRAME:033836/0823

Effective date: 20140912

AS Assignment

Owner name: MERRIMACK PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TANG, JIAN;REEL/FRAME:034259/0317

Effective date: 20141020

AS Assignment

Owner name: U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGEN

Free format text: SECURITY INTEREST;ASSIGNOR:MERRIMACK PHARMACEUTICALS, INC.;REEL/FRAME:037394/0285

Effective date: 20151222

AS Assignment

Owner name: MERRIMACK PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:U.S. BANK NATIONAL ASSOCIATION;REEL/FRAME:042254/0349

Effective date: 20170413

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION